Impact of oncogenic driver mutations on feedback between the PI3K and MEK pathways in cancer cells

Biosci Rep. 2012 Aug;32(4):413-22. doi: 10.1042/BSR20120050.

Abstract

Inhibition of the PI3K (phosphoinositide 3-kinase)/Akt/mTORC1 (mammalian target of rapamycin complex 1) and Ras/MEK [MAPK (mitogen-activated protein kinase)/ERK (extracellular-signal-regulated kinase) kinase]/ERK pathways for cancer therapy has been pursued for over a decade with limited success. Emerging data have indicated that only discrete subsets of cancer patients have favourable responses to these inhibitors. This is due to genetic mutations that confer drug insensitivity and compensatory mechanisms. Therefore understanding of the feedback mechanisms that occur with respect to specific genetic mutations may aid identification of novel biomarkers that predict patient response. In the present paper, we show that feedback between the PI3K/Akt/mTORC1 and Ras/MEK/ERK pathways is cell-line-specific and highly dependent on the activating mutation of K-Ras or overexpression c-Met. We found that cell lines exhibited differential signalling and apoptotic responses to PD184352, a specific MEK inhibitor, and PI103, a second-generation class I PI3K inhibitor. We reveal that feedback from the PI3K/Akt/mTORC1 to the Ras/MEK/ERK pathway is present in cancer cells harbouring either K-Ras activating mutations or amplification of c-Met but not the wild-type counterparts. Moreover, we demonstrate that inhibition of protein phosphatase activity by OA (okadaic acid) restored PI103-mediated feedback in wild-type cells. Together, our results demonstrate a novel mechanism for feedback between the PI3K/Akt/mTORC1 and the Ras/MEK/ERK pathways that only occurs in K-Ras mutant and c-Met amplified cells but not the isogenic wild-type cells through a mechanism that may involve inhibition of a specific endogenous phosphatase(s) activity. We conclude that monitoring K-Ras and c-Met status are important biomarkers for determining the efficacy of PI103 and other PI3K/Akt inhibitors in cancer therapy.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Apoptosis / drug effects
  • Benzamides / pharmacology
  • Cell Line, Tumor
  • Cell Survival / drug effects
  • Drug Synergism
  • Feedback, Physiological
  • Furans / pharmacology
  • Humans
  • MAP Kinase Signaling System*
  • Mechanistic Target of Rapamycin Complex 1
  • Mitogen-Activated Protein Kinases / antagonists & inhibitors
  • Mitogen-Activated Protein Kinases / metabolism
  • Multiprotein Complexes
  • Mutation*
  • Okadaic Acid / pharmacology
  • Oncogenes*
  • Phosphatidylinositol 3-Kinases / metabolism*
  • Phosphoinositide-3 Kinase Inhibitors
  • Phosphoprotein Phosphatases / antagonists & inhibitors
  • Phosphoprotein Phosphatases / metabolism
  • Phosphorylation
  • Protein Processing, Post-Translational
  • Proteins / antagonists & inhibitors
  • Proteins / metabolism
  • Proto-Oncogene Proteins / metabolism
  • Proto-Oncogene Proteins c-akt / metabolism
  • Proto-Oncogene Proteins c-met / genetics
  • Proto-Oncogene Proteins c-met / metabolism
  • Proto-Oncogene Proteins p21(ras)
  • Pyridines / pharmacology
  • Pyrimidines / pharmacology
  • Sirolimus / pharmacology
  • TOR Serine-Threonine Kinases
  • Up-Regulation
  • ras Proteins / metabolism

Substances

  • 2-(2-chloro-4-iodophenylamino)-N-cyclopropylmethoxy-3,4-difluorobenzamide
  • Benzamides
  • Furans
  • KRAS protein, human
  • Multiprotein Complexes
  • PI103
  • Phosphoinositide-3 Kinase Inhibitors
  • Proteins
  • Proto-Oncogene Proteins
  • Pyridines
  • Pyrimidines
  • Okadaic Acid
  • Proto-Oncogene Proteins c-met
  • Mechanistic Target of Rapamycin Complex 1
  • Proto-Oncogene Proteins c-akt
  • TOR Serine-Threonine Kinases
  • Mitogen-Activated Protein Kinases
  • Phosphoprotein Phosphatases
  • Proto-Oncogene Proteins p21(ras)
  • ras Proteins
  • Sirolimus